scispace - formally typeset
Search or ask a question
Author

Kyle J. Travaglini

Bio: Kyle J. Travaglini is an academic researcher from Stanford University. The author has contributed to research in topics: Cell type & Biology. The author has an hindex of 9, co-authored 11 publications receiving 942 citations. Previous affiliations of Kyle J. Travaglini include University of California, Los Angeles.
Topics: Cell type, Biology, Cell, Atlas (anatomy), Medicine

Papers
More filters
Journal ArticleDOI
18 Nov 2020-Nature
TL;DR: Droplet- and plate-based single cell RNA sequencing applied to ~75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, have allowed them to define the gene expression profiles and anatomical locations of 58 cell populations in the human lung.
Abstract: Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1–9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution. Expression profiling on 75,000 single cells creates a comprehensive cell atlas of the human lung that includes 41 out of 45 previously known cell types and 14 new ones.

795 citations

Journal ArticleDOI
Nicole Almanzar, Jane Antony, Ankit S. Baghel, Isaac Bakerman, Ishita Bansal, Ben A. Barres, Philip A. Beachy, Daniela Berdnik, Biter Bilen, Douglas Brownfield, Corey Cain, Charles Chan, Michelle B. Chen, Michael F. Clarke, Stephanie D. Conley, Spyros Darmanis, Aaron Demers, Kubilay Demir, Antoine de Morrée, Tessa Divita, Haley du Bois, Hamid Ebadi, F. Hernan Espinoza, Matt Fish, Qiang Gan, Benson M. George, Astrid Gillich, Rafael Gòmez-Sjöberg, Foad Green, Geraldine Genetiano, Xueying Gu, Gunsagar S. Gulati, Oliver Hahn, Michael S. Haney, Yan Hang, Lincoln Harris, Mu He, Shayan Hosseinzadeh, Albin Huang, Kerwyn Casey Huang, Tal Iram, Taichi Isobe, Feather Ives, Robert C. Jones, Kevin S. Kao, Jim Karkanias, Guruswamy Karnam, Andreas Keller, Aaron M. Kershner, Nathalie Khoury, Seung K. Kim, Bernhard M. Kiss, William Kong, Mark A. Krasnow, Maya E. Kumar, Christin S. Kuo, Jonathan K. Lam, Davis P. Lee, Song E. Lee, Benoit Lehallier, Olivia Leventhal, Guang Li, Qingyun Li, Ling Liu, Annie Lo, Wan Jin Lu, Maria F. Lugo-Fagundo, Anoop Manjunath, Andrew May, Ashley Maynard, Aaron McGeever, Marina McKay, M. Windy McNerney, Bryan D. Merrill, Ross J. Metzger, Marco Mignardi, Dullei Min, Ahmad N. Nabhan, Norma Neff, Katharine M. Ng, Patricia K. Nguyen, Joseph Noh, Roel Nusse, Róbert Pálovics, Rasika Patkar, Weng Chuan Peng, Lolita Penland, Angela Oliveira Pisco, Katherine S. Pollard, Robert Puccinelli, Zhen Qi, Stephen R. Quake, Thomas A. Rando, Eric J. Rulifson, Nicholas Schaum, Joe M. Segal, Shaheen S. Sikandar, Rahul Sinha, Rene V. Sit, Justin L. Sonnenburg, Daniel Staehli, Krzysztof Szade, Michelle Tan, Weilun Tan, Cristina M. Tato, Krissie Tellez, Laughing Bear Torrez Dulgeroff, Kyle J. Travaglini, Carolina Tropini, Margaret Tsui, Lucas M. Waldburger, Bruce Wang, Linda J. van Weele, Kenneth I. Weinberg, Irving L. Weissman, Michael N. Wosczyna, Sean M. Wu, Tony Wyss-Coray, Jinyi Xiang, Soso Xue, Kevin A. Yamauchi, Andrew C. Yang, Lakshmi P. Yerra, Justin Youngyunpipatkul, Brian Yu, Fabio Zanini, Macy E. Zardeneta, Alexander Zee, Chunyu Zhao, Fan Zhang, Hui Zhang, Martin J. Zhang, Lu Zhou, James Zou 
23 Jul 2020-Nature

517 citations

Posted ContentDOI
Pascal Barbry1, Christoph Muus2, Christoph Muus3, Malte D Luecken, Gökcen Eraslan3, Avinash Waghray2, Graham Heimberg3, Lisa Sikkema, Yoshihiko Kobayashi4, Eeshit Dhaval Vaishnav5, Ayshwarya Subramanian3, Christopher Smilie3, Karthik A. Jagadeesh3, Elizabeth Thu Duong6, Evgenij Fiskin3, Elena Torlai Triglia3, Meshal Ansari, Peiwen Cai7, Brian M. Lin2, Justin Buchanan6, Sijia Chen8, Jian Shu5, Jian Shu3, Adam L. Haber2, Adam L. Haber3, Hattie Chung3, Daniel T. Montoro3, Taylor Adams9, Hananeh Aliee, J. Samuel10, Allon Zaneta Andrusivova11, Ilias Angelidis, Orr Ashenberg3, Kevin Bassler12, Christophe Bécavin1, Inbal Benhar2, Joseph Bergenstråhle11, Ludvig Bergenstråhle11, Liam Bolt13, Emelie Braun14, Linh T. Bui15, Mark Chaffin3, Evgeny Chichelnitskiy16, Joshua Chiou6, Thomas M. Conlon, Michael S. Cuoco3, Marie Deprez1, David Fischer, Astrid Gillich, Joshua Gould3, Minzhe Guo17, Austin J. Gutierrez15, Arun C. Habermann18, Tyler Harvey3, Peng He13, Xiaomeng Hou6, Xiaomeng Hou8, Lijuan Hu14, Alok Jaiswal3, Peiyong Jiang19, Theodoros Kapellos12, Christin S. Kuo, Ludvig Larsson11, Michael Leney-Greene3, Kyungtae Lim20, Monika Litviňuková21, Monika Litviňuková13, Ji Lu19, Leif S. Ludwig3, Wendy Luo3, Henrike Maatz21, Elo Madissoon13, Lira Mamanova13, Kasidet Manakongtreecheep3, Kasidet Manakongtreecheep2, Charles-Hugo Marquette1, Ian Mbano, Alexi McAdams22, Ross J. Metzger, Ahmad N. Nabhan, Sarah K. Nyquist10, Lolita Penland, Olivier Poirion6, Sergio Poli9, Cancan Qi23, Rachel Queen24, Daniel Reichart2, Daniel Reichart25, Ivan O. Rosas9, Jonas C. Schupp9, Rahul Sinha, Rene Sit, Kamil Slowikowski2, Kamil Slowikowski3, Michal Slyper3, Neal Smith3, Neal Smith2, Alex Sountoulidis26, Maximilian Strunz, Dawei Sun20, Carlos Talavera-López13, Peng Tan3, Jessica Tantivit3, Jessica Tantivit2, Kyle J. Travaglini, Nathan R. Tucker3, Katherine A. Vernon3, Katherine A. Vernon8, Marc Wadsworth10, Julia Waldman3, Xiuting Wang7, Wenjun Yan2, William Zhao7, Carly Ziegler10 
20 Apr 2020-bioRxiv
TL;DR: Differences in the cell type-specific expression of mediators of SARS-CoV-2 viral entry may be responsible for aspects of COVID-19 epidemiology and clinical course, and point to putative molecular pathways involved in disease susceptibility and pathogenesis.
Abstract: The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, creates an urgent need for identifying molecular mechanisms that mediate viral entry, propagation, and tissue pathology. Cell membrane bound angiotensin-converting enzyme 2 (ACE2) and associated proteases, transmembrane protease serine 2 (TMPRSS2) and Cathepsin L (CTSL), were previously identified as mediators of SARS-CoV2 cellular entry. Here, we assess the cell type-specific RNA expression of ACE2 , TMPRSS2 , and CTSL through an integrated analysis of 107 single-cell and single-nucleus RNA-Seq studies, including 22 lung and airways datasets (16 unpublished), and 85 datasets from other diverse organs. Joint expression of ACE2 and the accessory proteases identifies specific subsets of respiratory epithelial cells as putative targets of viral infection in the nasal passages, airways, and alveoli. Cells that co-express ACE2 and proteases are also identified in cells from other organs, some of which have been associated with COVID-19 transmission or pathology, including gut enterocytes, corneal epithelial cells, cardiomyocytes, heart pericytes, olfactory sustentacular cells, and renal epithelial cells. Performing the first meta-analyses of scRNA-seq studies, we analyzed 1,176,683 cells from 282 nasal, airway, and lung parenchyma samples from 164 donors spanning fetal, childhood, adult, and elderly age groups, associate increased levels of ACE2 , TMPRSS2 , and CTSL in specific cell types with increasing age, male gender, and smoking, all of which are epidemiologically linked to COVID-19 susceptibility and outcomes. Notably, there was a particularly low expression of ACE2 in the few young pediatric samples in the analysis. Further analysis reveals a gene expression program shared by ACE2 + TMPRSS2 + cells in nasal, lung and gut tissues, including genes that may mediate viral entry, subtend key immune functions, and mediate epithelial-macrophage cross-talk. Amongst these are IL6, its receptor and co-receptor, IL1R , TNF response pathways, and complement genes. Cell type specificity in the lung and airways and smoking effects were conserved in mice. Our analyses suggest that differences in the cell type-specific expression of mediators of SARS-CoV-2 viral entry may be responsible for aspects of COVID-19 epidemiology and clinical course, and point to putative molecular pathways involved in disease susceptibility and pathogenesis.

244 citations

Journal ArticleDOI
14 Oct 2020-Nature
TL;DR: Single-cell analysis of blood vessels in the alveolus, the site of chronic disease and virus-induced lung injury, reveals two intermingled endothelial cell types with specialized gas exchange and stem cell functions.
Abstract: In the mammalian lung, an apparently homogenous mesh of capillary vessels surrounds each alveolus, forming the vast respiratory surface across which oxygen transfers to the blood1. Here we use single-cell analysis to elucidate the cell types, development, renewal and evolution of the alveolar capillary endothelium. We show that alveolar capillaries are mosaics; similar to the epithelium that lines the alveolus, the alveolar endothelium is made up of two intermingled cell types, with complex 'Swiss-cheese'-like morphologies and distinct functions. The first cell type, which we term the 'aerocyte', is specialized for gas exchange and the trafficking of leukocytes, and is unique to the lung. The other cell type, termed gCap ('general' capillary), is specialized to regulate vasomotor tone, and functions as a stem/progenitor cell in capillary homeostasis and repair. The two cell types develop from bipotent progenitors, mature gradually and are affected differently in disease and during ageing. This cell-type specialization is conserved between mouse and human lungs but is not found in alligator or turtle lungs, suggesting it arose during the evolution of the mammalian lung. The discovery of cell type specialization in alveolar capillaries transforms our understanding of the structure, function, regulation and maintenance of the air-blood barrier and gas exchange in health, disease and evolution.

178 citations

Posted ContentDOI
27 Aug 2019-bioRxiv
TL;DR: This comprehensive molecular atlas elucidates the biochemical functions of lung cell types and the cell-selective transcription factors and optimal markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signaling interactions; and identifies the cell types directly affected by lung disease genes.
Abstract: Although single cell RNA sequencing studies have begun providing compendia of cell expression profiles, it has proven more difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here we describe droplet- and plate-based single cell RNA sequencing applied to ∼70,000 human lung and blood cells, combined with a multi-pronged cell annotation approach, which have allowed us to define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 of 45 previously known cell types or subtypes and 14 new ones. This comprehensive molecular atlas elucidates the biochemical functions of lung cell types and the cell-selective transcription factors and optimal markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signaling interactions including sources and targets of chemokines in immune cell trafficking and expression changes on lung homing; and identifies the cell types directly affected by lung disease genes. Comparison to mouse identified 17 molecular types that appear to have been gained or lost during lung evolution and others whose expression profiles have been substantially altered, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This lung atlas provides the molecular foundation for investigating how lung cell identities, functions, and interactions are achieved in development and tissue engineering and altered in disease and evolution.

100 citations


Cited by
More filters
01 Jan 2020
TL;DR: Prolonged viral shedding provides the rationale for a strategy of isolation of infected patients and optimal antiviral interventions in the future.
Abstract: Summary Background Since December, 2019, Wuhan, China, has experienced an outbreak of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Epidemiological and clinical characteristics of patients with COVID-19 have been reported but risk factors for mortality and a detailed clinical course of illness, including viral shedding, have not been well described. Methods In this retrospective, multicentre cohort study, we included all adult inpatients (≥18 years old) with laboratory-confirmed COVID-19 from Jinyintan Hospital and Wuhan Pulmonary Hospital (Wuhan, China) who had been discharged or had died by Jan 31, 2020. Demographic, clinical, treatment, and laboratory data, including serial samples for viral RNA detection, were extracted from electronic medical records and compared between survivors and non-survivors. We used univariable and multivariable logistic regression methods to explore the risk factors associated with in-hospital death. Findings 191 patients (135 from Jinyintan Hospital and 56 from Wuhan Pulmonary Hospital) were included in this study, of whom 137 were discharged and 54 died in hospital. 91 (48%) patients had a comorbidity, with hypertension being the most common (58 [30%] patients), followed by diabetes (36 [19%] patients) and coronary heart disease (15 [8%] patients). Multivariable regression showed increasing odds of in-hospital death associated with older age (odds ratio 1·10, 95% CI 1·03–1·17, per year increase; p=0·0043), higher Sequential Organ Failure Assessment (SOFA) score (5·65, 2·61–12·23; p Interpretation The potential risk factors of older age, high SOFA score, and d-dimer greater than 1 μg/mL could help clinicians to identify patients with poor prognosis at an early stage. Prolonged viral shedding provides the rationale for a strategy of isolation of infected patients and optimal antiviral interventions in the future. Funding Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences; National Science Grant for Distinguished Young Scholars; National Key Research and Development Program of China; The Beijing Science and Technology Project; and Major Projects of National Science and Technology on New Drug Creation and Development.

4,408 citations

Posted ContentDOI
02 Nov 2018-bioRxiv
TL;DR: This work presents a strategy for comprehensive integration of single cell data, including the assembly of harmonized references, and the transfer of information across datasets, and demonstrates how anchoring can harmonize in-situ gene expression and scRNA-seq datasets.
Abstract: Single cell transcriptomics (scRNA-seq) has transformed our ability to discover and annotate cell types and states, but deep biological understanding requires more than a taxonomic listing of clusters. As new methods arise to measure distinct cellular modalities, including high-dimensional immunophenotypes, chromatin accessibility, and spatial positioning, a key analytical challenge is to integrate these datasets into a harmonized atlas that can be used to better understand cellular identity and function. Here, we develop a computational strategy to "anchor" diverse datasets together, enabling us to integrate and compare single cell measurements not only across scRNA-seq technologies, but different modalities as well. After demonstrating substantial improvement over existing methods for data integration, we anchor scRNA-seq experiments with scATAC-seq datasets to explore chromatin differences in closely related interneuron subsets, and project single cell protein measurements onto a human bone marrow atlas to annotate and characterize lymphocyte populations. Lastly, we demonstrate how anchoring can harmonize in-situ gene expression and scRNA-seq datasets, allowing for the transcriptome-wide imputation of spatial gene expression patterns, and the identification of spatial relationships between mapped cell types in the visual cortex. Our work presents a strategy for comprehensive integration of single cell data, including the assembly of harmonized references, and the transfer of information across datasets. Availability: Installation instructions, documentation, and tutorials are available at: https://www.satijalab.org/seurat

2,037 citations

DOI
01 Jan 2020

1,967 citations

Journal ArticleDOI
TL;DR: It is proposed that the Pearson residuals from “regularized negative binomial regression,” where cellular sequencing depth is utilized as a covariate in a generalized linear model, successfully remove the influence of technical characteristics from downstream analyses while preserving biological heterogeneity.
Abstract: Single-cell RNA-seq (scRNA-seq) data exhibits significant cell-to-cell variation due to technical factors, including the number of molecules detected in each cell, which can confound biological heterogeneity with technical effects. To address this, we present a modeling framework for the normalization and variance stabilization of molecular count data from scRNA-seq experiments. We propose that the Pearson residuals from “regularized negative binomial regression,” where cellular sequencing depth is utilized as a covariate in a generalized linear model, successfully remove the influence of technical characteristics from downstream analyses while preserving biological heterogeneity. Importantly, we show that an unconstrained negative binomial model may overfit scRNA-seq data, and overcome this by pooling information across genes with similar abundances to obtain stable parameter estimates. Our procedure omits the need for heuristic steps including pseudocount addition or log-transformation and improves common downstream analytical tasks such as variable gene selection, dimensional reduction, and differential expression. Our approach can be applied to any UMI-based scRNA-seq dataset and is freely available as part of the R package sctransform, with a direct interface to our single-cell toolkit Seurat.

1,898 citations

Journal ArticleDOI
Nicolas Vabret1, Graham J. Britton1, Conor Gruber1, Samarth Hegde1, Joel Kim1, Maria Kuksin1, Rachel Levantovsky1, Louise Malle1, Alvaro Moreira1, Matthew D. Park1, Luisanna Pia1, Emma Risson1, Miriam Saffern1, Bérengère Salomé1, Myvizhi Esai Selvan1, Matthew P. Spindler1, Jessica Tan1, Verena van der Heide1, Jill Gregory1, Konstantina Alexandropoulos1, Nina Bhardwaj1, Brian D. Brown1, Benjamin Greenbaum1, Zeynep H. Gümüş1, Dirk Homann1, Amir Horowitz1, Alice O. Kamphorst1, Maria A. Curotto de Lafaille1, Saurabh Mehandru1, Miriam Merad1, Robert M. Samstein1, Manasi Agrawal, Mark Aleynick, Meriem Belabed, Matthew Brown1, Maria Casanova-Acebes, Jovani Catalan, Monica Centa, Andrew Charap, Andrew K Chan, Steven T. Chen, Jonathan Chung, Cansu Cimen Bozkus, Evan Cody, Francesca Cossarini, Erica Dalla, Nicolas F. Fernandez, John A. Grout, Dan Fu Ruan, Pauline Hamon, Etienne Humblin, Divya Jha, Julia Kodysh, Andrew Leader, Matthew Lin, Katherine E. Lindblad, Daniel Lozano-Ojalvo, Gabrielle Lubitz, Assaf Magen, Zafar Mahmood2, Gustavo Martinez-Delgado, Jaime Mateus-Tique, Elliot Meritt, Chang Moon1, Justine Noel, Timothy O'Donnell, Miyo Ota, Tamar Plitt, Venu Pothula, Jamie Redes, Ivan Reyes Torres, Mark P. Roberto, Alfonso R. Sanchez-Paulete, Joan Shang, Alessandra Soares Schanoski, Maria Suprun, Michelle Tran, Natalie Vaninov, C. Matthias Wilk, Julio A. Aguirre-Ghiso, Dusan Bogunovic1, Judy H. Cho, Jeremiah J. Faith, Emilie K. Grasset, Peter S. Heeger, Ephraim Kenigsberg, Florian Krammer1, Uri Laserson1 
16 Jun 2020-Immunity
TL;DR: The current state of knowledge of innate and adaptive immune responses elicited by SARS-CoV-2 infection and the immunological pathways that likely contribute to disease severity and death are summarized.

1,350 citations